Context.—

Excision repair cross-complementation 1 (ERCC1) is a key enzyme in nuclear excision repair pathway and has a critical role in helping remove DNA adducts caused by cross-linking agents, such as platinum-containing cancer chemotherapies and other DNA-damaging therapeutic modalities. ERCC1 expression, evaluated by techniques such as immunohistochemistry, has been associated with clinical response; ERCC1+ tumors are more resistant to cisplatin treatment than are ERCC1 tumors. Although several immunohistochemistry, anti-ERCC1 antibodies are available, the 8F1 clone, in particular, has been used in many studies. Recent evidence has suggested that the 8F1 antibody cross-reacts with at least one other protein, raising concerns about the specificity of this clone.

Objective.—

To design an immunohistochemistry assay to detect ERCC1 levels that show dynamic range and consistent analytic performance.

Design.—

Two different primary antibodies to ERCC1, clones 4F9 and D6G6, were evaluated on formalin-fixed, paraffin-embedded tissue. We then performed a fit-for-purpose assay validation with the 4F9 clone, which included sensitivity assessment across several solid tumor types and evaluation of analytic parameters, such as precision and reproducibility.

Results.—

The 4F9 clone was consistently superior to the D6G6 clone in the optimization phase. A range of expression was seen in ovarian, head and neck, non–small cell lung, and esophageal cancer samples when tested with the 4F9 clone. The antibody showed acceptable reproducibility (31.02%) and precision (16.06%).

Conclusions.—

This assay can be used to assess ERCC1 levels during clinical studies of patient tumors from a variety of tumor types.

Platinum-based therapies, such as cisplatin, carboplatin, and oxaliplatin, are standard chemotherapeutic agents that are used to treat different tumor types, including non–small cell lung cancer (NSCLC), head and neck cancer, gastric cancer, bladder cancer, and colorectal cancer, among others.13  Platinum compounds react with DNA to form adducts. These adducts consist of platinum-DNA monoadducts, intrastrand and interstrand cross-links, and/or DNA-protein cross-links.4  These DNA lesions can be repaired by the nucleotide excision repair pathway.1,2,5 

Within the nucleotide excision repair pathway, the excision repair cross-complementing group 1 protein (ERCC1) has a critical and rate-limiting role of recognizing and helping remove DNA adducts.5  Specifically, ERCC1 forms a heterodimer with ERCC4 (previously XPF) and incises the 5′ strand of damaged DNA.1,2  Four isoforms of the ERCC1 protein (201, 202, 203, and 204) are generated by alternative splicing of the ERCC1 gene.6  Mutations in the 201 and 203 isoforms disrupt the ERCC1 capacity in the nucleotide excision repair pathway, suggesting that these are the isoforms responsible for DNA repair.6  As a biomarker, ERCC1 expression in tumors assessed by immunohistochemistry (IHC), Western blot, and reverse transcription-polymerase chain reaction is potentially predictive of response to platinum-based adjuvant chemotherapy in a variety of cancers, including lung and ovarian cancer.1  Many studies have shown the correlation of high ERCC1 expression and poor response to therapy across different tumor types.5,715  In most of these studies, the antibody used to assess ERCC1 expression has been the monoclonal 8F1 antibody.

Recent evidence has raised questions about the specificity of the 8F1 antibody for ERCC1.6,7,16  The 8F1 antibody not only recognizes ERCC1 but also recognizes a nuclear membrane protein PCYT1A.6,1618  This nonspecificity and potential lot-to-lot difference in the 8F1 antibody have cast doubts both on the utility of this antibody and on some of the data that has shown ERCC1 protein expression being predictive for platinum-based therapies in solid tumors.6 

In addition to 8F1, there are several alternative ERCC1 antibodies available for IHC purposes. One antibody that has been used for IHC is the 4F9 clone. A direct comparison between the 4F9 clone and the 8F1 clone has shown that the 4F9 clone was more specific and did not cross-react with other proteins.16  This antibody has also been used to develop an ERCC1 assay specific for colorectal tumors.4  Additionally, some groups have examined the isoforms targeted by the 4F9 epitope to confirm that the isoforms relevant for DNA repair were included.4  The development of an antibody that would target only the isoforms responsible for DNA repair is currently a high priority for researchers interested in ERCC1; however, because none of the isoforms have isoform-specific sequences, that is challenging.6 

The objective of this study was to test the performance of 2 anti-ERCC1 antibody clones, 4F9 and D6G6, to develop and validate an ERCC1-specific IHC assay for formalin-fixed, paraffin-embedded tissue (FFPE) that could be used for clinical trial samples.

Cell Line and Tumor Tissue Samples

LUDLU-1 and NCI-H520 cells were selected as medium- and high-expressing cells, respectively, based on internal Western blot data (anti-ERCC1, AC-0206RUO; Epitomics, Burlingame, California). Cells were fixed in formalin and paraffin embedded. These cell-pellet blocks were used as run controls for the duration of the validation. Human tonsil and a breast cancer sample were used as tissue controls. One hundred twenty-five FFPE processed, surgically resected human tumor blocks were purchased commercially for use in this study (Asterand BioScience, Detroit, Michigan; ProteoGenex, Culver City, California). Tumor types included NSCLC (squamous cell and adenocarcinoma subtypes as 2 distinct categories), head and neck, esophageal, and ovarian cancer of varying histologic subtypes (Table 1). Additionally, FFPE blocks from 31 different healthy tissues were used to assess assay specificity.

Table 1. 

Commercially Procured Tumor-Specimen Histology and Sex Data

Commercially Procured Tumor-Specimen Histology and Sex Data
Commercially Procured Tumor-Specimen Histology and Sex Data

Immunohistochemistry

Formalin-fixed, paraffin-embedded tissue samples were sectioned at 4 to 5 μm and placed onto positively charged glass slides. Sections were deparaffinized, rehydrated, and heat treated in a pressurized decloaking chamber (DC2008US; Biocare Medical, Concord, California) in EDTA buffer at 125°C, for 30 seconds and then cooled for 15 minutes to 95°C, for antigen retrieval. Slides were cooled to room temperature and incubated in a peroxidase blocking solution (Dako, Carpinteria, California) for 10 minutes. Slides were washed and incubated with the primary antibodies (ERCC1 and 4F9, OriGene, Rockville, Maryland; ERCC1 and D6G6, Cell Signaling Technology, Danvers, Massachusetts) for 1 hour at room temperature. Several iterations of antigen retrieval, primary antibody dilution, and incubation times were tested to determine the optimal conditions (Table 2). Simultaneously, a negative control-isotype slide was incubated with Universal Negative Mouse antibody (Dako) for 1 hour at room temperature. Detection was performed by incubation with polymer-horseradish peroxidase reagent (Dako) for 30 minutes, followed by incubated with 3,3′-diaminobenzidine chromogen for 5 minutes. Slides were counterstained with hematoxylin and coverslipped before being scanned using the iScan Coreo slide scanner (Ventana, Tucson, Arizona). All staining was performed manually.

Table 2. 

Optimization Conditions Testeda

Optimization Conditions Testeda
Optimization Conditions Testeda

ERCC1 Assessment and Pathology Scoring

The scoring system included a semiquantitative analysis of nuclear staining intensity. The staining intensity of ERCC1 was judged relative to the intensity of the internal stromal fibroblasts and/or endothelial cells as well as staining of the negative-control antibody. Internal positive controls immediately adjacent to the tumor area had to show positive staining for samples to be scored. Staining in the stromal cells was considered 2+, and the staining intensity of the tumor cells was scored in comparison to the level of 2+ exhibited in those adjacent stromal cells. The study pathologist (David W. Rogers, MD, LabCorp Clinical Trials) scored the samples by taking a minimum of 3 to 4 fields of view at the appropriate magnification (×40) and analyzed regions in which tumor and immediate adjacent stromal cells showed comparable reactivity. Outputs included both the percentage of positive cell results and an H score, a semiquantitative score derived by multiplying the percentage of the tumor at each intensity bin by the intensity number. This is illustrated in the formula below:

Similar to previous studies, the median H score per tumor type was used as a cutoff to define tumors ERCC1 positive or negative results.4,6 A sample was considered ERCC1+ if the above internal control criteria were met and the assigned H score was greater than or equal to the median of the H scores for that specific tumor type. Negative staining of adjacent stromal cells resulted in a sample being scored not evaluable, regardless of tumor cell staining seen in the sample.

Antibody Screening and Assay Optimization

Initial screening was performed with 2 primary antibodies to ERCC1, clones 4F9 and D6G6, and a separate assay for each clone was optimized and developed (Table 2). Testing was done on FFPE blocks of 2 human tonsil cancers, 1 breast carcinoma, and a cell line with relatively high ERCC1 expression, NCI-520, based on Western blot analysis (data not shown). After optimization of each antibody, 25 FFPE NSCLC adenocarcinoma samples were stained with each antibody. Figure 1, A and B, shows ERCC1 staining with clone 4F9 and D6G6, respectively. After careful evaluation, it was deemed that the 4F9 clone provided a good signal to noise ratio and overall better ERCC1 staining in both cell-line controls and NSCLC adenocarcinoma samples (Figure 1). Even at high concentrations, the D6G6 clone staining was weak in all samples stained. From this evaluation, the 4F9 clone was moved forward for further evaluation and assay validation.

Figure 1. 

Excision repair cross-complementation group 1 protein clone comparison in non–small cell lung cancer adenocarcinoma sample (4F9 clone, original magnification ×20 [A]; D6G6 clone, original magnification ×20 [B]).

Figure 1. 

Excision repair cross-complementation group 1 protein clone comparison in non–small cell lung cancer adenocarcinoma sample (4F9 clone, original magnification ×20 [A]; D6G6 clone, original magnification ×20 [B]).

Close modal

ERCC1 Expression in Human Tumors

To better understand the ERCC1 expression across various solid tumors in which platinum therapy could be used for chemotherapy, we evaluated NSCLC, esophageal, head and neck, and ovarian tumor samples for ERCC1 expression using the 4F9 clone (Figure 2, A through E). Then, 125 surgically resected, FFPE-processed tumors were tested. The sample breakdown within each tumor type is listed in Table 3. From an initial cohort of 125 samples, 102 (82%) were deemed ERCC1 evaluable because of sufficient staining in the fibroblasts, and/or endothelial or stromal cells (Figure 3). The ERCC1+ and ERCC1 samples for each tumor type is shown in Table 3. Overall, among the tumors tested, the frequency of ERCC1 positivity was similar to what has been previously reported (Table 4). There was no difference in staining patterns among the various tumor types.

Figure 2. 

Excision repair cross-complementation group 1 protein (ERCC1)–positive staining in various tumor types. All 5 tumor types tested included samples that showed ERCC1 positivity. Samples tested included (A) non small cell lung cancer (NSCLC) adenocarcinoma, (B) NSCLC squamous cell carcinoma, (C) esophageal cancer, (D) head and neck cancer, and (E) ovarian cancer (original magnification ×20).

Figure 2. 

Excision repair cross-complementation group 1 protein (ERCC1)–positive staining in various tumor types. All 5 tumor types tested included samples that showed ERCC1 positivity. Samples tested included (A) non small cell lung cancer (NSCLC) adenocarcinoma, (B) NSCLC squamous cell carcinoma, (C) esophageal cancer, (D) head and neck cancer, and (E) ovarian cancer (original magnification ×20).

Close modal
Table 3. 

Summary of Excision Repair Cross-Complementation Group 1 Protein (ERCC1) Results Across Tumor Types

Summary of Excision Repair Cross-Complementation Group 1 Protein (ERCC1) Results Across Tumor Types
Summary of Excision Repair Cross-Complementation Group 1 Protein (ERCC1) Results Across Tumor Types
Figure 3. 

H score by tumor type. ERCC1 H scores for non–small cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cell carcinoma, esophageal cancer, head and neck cancer, and ovarian samples.

Figure 3. 

H score by tumor type. ERCC1 H scores for non–small cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cell carcinoma, esophageal cancer, head and neck cancer, and ovarian samples.

Close modal
Table 4. 

Literature Review of Previously Published Data on Excision Repair Cross-Complementation Group 1 Protein (ERCC1) Status

Literature Review of Previously Published Data on Excision Repair Cross-Complementation Group 1 Protein (ERCC1) Status
Literature Review of Previously Published Data on Excision Repair Cross-Complementation Group 1 Protein (ERCC1) Status

Analytic Validation

Precision and reproducibility analyses were performed to determine the intra-assay and interassay variability, respectively, for the ERCC1 assay. Samples used here included the controls used in the optimization phase and a series of various tumor types that had been characterized for ERCC1 and that showed a range of expressions based on the H score (Figure 3). To assess assay precision, 5 serial sections from 12 tumor blocks and 5 controls were stained on one day. Similarly, to assess reproducibility, a slide from 11 tumor blocks and 5 controls was cut and stained on 5 different days. A mixed-effect model was used to obtain variance components for precision and reproducibility. In the mixed-effect model, variation in replicated runs from the same sample was treated as a random effect. The coefficient of variation (CV) for precision and reproducibility was derived from variance components for precision and reproducibility, respectively. Overall, we found the ERCC1 assay CV for precision (16.06%) and reproducibility (31.02%) to be acceptable (Table 3; Figures 4 and 5).

Figure 4. 

Precision results from H score analysis. Replicates stained in the same run. The following samples were used for the precision analysis: non–small cell lung cancer (NSCLC) adenocarcinoma (n = 5; 29%), NSCLC squamous cell carcinoma (n = 3; 18%), esophageal cancer (n = 4; 24%), healthy tonsil tissue (n = 2; 12%), breast cancer (n = 1; 6%), and cell lines (n = 2; 12%).

Figure 4. 

Precision results from H score analysis. Replicates stained in the same run. The following samples were used for the precision analysis: non–small cell lung cancer (NSCLC) adenocarcinoma (n = 5; 29%), NSCLC squamous cell carcinoma (n = 3; 18%), esophageal cancer (n = 4; 24%), healthy tonsil tissue (n = 2; 12%), breast cancer (n = 1; 6%), and cell lines (n = 2; 12%).

Close modal
Figure 5. 

Reproducibility results from H score analysis. Replicates stained on different days. The following samples were used in the reproducibility analysis: non–small cell lung cancer (NSCLC) adenocarcinoma (n = 4; 25%), NSCLC squamous cell carcinoma (n = 3; 19%), esophageal cancer (n = 4, 25%), healthy tonsil tissue (n = 2; 13%), breast cancer (n = 1; 6%), and cell lines (n = 2; 13%).

Figure 5. 

Reproducibility results from H score analysis. Replicates stained on different days. The following samples were used in the reproducibility analysis: non–small cell lung cancer (NSCLC) adenocarcinoma (n = 4; 25%), NSCLC squamous cell carcinoma (n = 3; 19%), esophageal cancer (n = 4, 25%), healthy tonsil tissue (n = 2; 13%), breast cancer (n = 1; 6%), and cell lines (n = 2; 13%).

Close modal

Specificity of the ERCC1 assay was assessed by testing the assay on a panel of normal human tissues. The assay exhibited minimal positive staining in this panel of tissues, with no unexpected findings. Of 93 samples tested, 3 (3.2%) showed positivity (H score ≥ 160, median of entire data set), and there was no obvious endothelium expression observed in healthy tissues (Table 5).

Table 5. 

Specificity Samples Tested

Specificity Samples Tested
Specificity Samples Tested

In this study, we performed a fit-for-purpose validation of an ERCC1 IHC assay that can be used to analyze samples in an exploratory setting. We found that the 4F9 clone provided the best ERCC1 signal, with no nonspecific staining among the 2 antibodies tested. We validated this ERCC1 assay using a variety of tumors types, including NSCLC, esophageal, head and neck, and ovarian cancer. The sensitivity analysis using this assay showed that there was a wide range of ERCC1 expression within the various tumor types (Table 4). This is consistent with previously published data that has shown that ERCC1-high and ERCC1-low populations existed within tumor types. Other groups have recently explored the use of this antibody clone to characterize the levels of ERCC1 in head and neck cancer as well as colorectal cancer.4,18 

As part of the assay validation, we further evaluated parameters including precision, reproducibility, and specificity to ensure that the assay met the fit-for-purpose criteria. Published guidelines from the American Society of Clinical Oncology and the College of American Pathologists offer an in-depth literature review, coupled with recommendations to improve IHC assay specificity for established markers, such as HER2 across laboratories.19,20  The College of American Pathologists Pathology and Laboratory Quality Center have provided recent information for the analytic validation of IHC assays meant to establish a novel assay or compare a novel assay to an existing assay.21  Bioanalytic assays, such as enzyme-linked immunosorbent assays, ligand-binding assays, or pharmacokinetic assays to measure drug exposure, have strict coefficient of variation acceptance levels in which assays should not exceed more than 20% for parameters such as precision and reproducibility during the validation.22  These parameters are noted and recommended for testing in both the recent American Society of Clinical Oncology and College of American Pathologists publications, but strict acceptance criteria for CVs are not standard for IHC assays. Here, for the analytic testing, we used the same tumor sample set to measure precision and reproducibility. After thorough testing of both precision and reproducibility, we found that the CVs were low, 16.06% and 31.02%, respectively. In our experience, when validating IHC assays, higher CVs are observed in reproducibility testing than in precision; this can be attributed, in part, to reproducibility testing being performed to test the day-to-day variability, whereas precision testing is performed in one run.

In this study, we tested tumor samples that were procured from commercial sources. The advantage of using commercial sources is that tumor tissue can be collected at a reasonable cost and assay validations can be performed quickly. There are some inherent limitations with commercial tumor samples, including the inability to perform analyses, such as correlating ERCC1 protein levels and response because of a lack of available patient treatment and follow-up data. Additionally, these samples came from multiple clinical sites, and information about fixation time and processing methods was unavailable. However, the potential variety in tissue processing (e.g., fixation time) for these types of samples could mimic the variety of sample preparations seen when collecting samples from different clinical sites in a global study. In this study, the cell-line controls were generated under controlled fixation and processing conditions.

The nature of our scoring algorithm is similar to previous publications looking at ERCC1 expression, and potential stratification methods could include dividing groups into positive and negative results based on median or mean ERCC1 expression. The scoring here was based, in part, on the tumor tissue containing internal normal elements and the staining (intensity) of these structures when calculating the ERCC1 expression in the tumor area. A subset of tumor samples tested (n = 23, 18.4%) had no staining in fibroblasts or endothelial cells and were thus deemed not evaluable. Although the internal stromal controls may make a more stringent cutoff of the data, the staining and analysis of these normal elements allows for the control of differences in fixation.7  In potential clinical studies that would use this assay prospectively, because of the strict scoring criteria of internal-control staining, the screen failure rate would have to be monitored. In situations in which internal normal elements do not stain, testing fresh tumor biopsies could be an option to diminish the effects potentially caused by poor tissue processing.

This assay is now a validated, laboratory-developed test and can be used to measure ERCC1 protein levels in clinical trial–related FFPE samples. Transfer of this assay to another laboratory would require a bridge study that meets the concordance guidelines from the American Society of Clinical Oncology and the College of American Pathologists.19 

We thank R. Smale, BS, and D. W. Rogers, MD, for their assistance in staining and analyzing the samples. We thank E. Lightcap, PhD, for providing Western blot data for ERCC1 on a panel of cell lines.

1
Bowden
NA.
Nucleotide excision repair: why is it not used to predict response to platinum-based chemotherapy?
Cancer Lett
.
2014
;
346
(
2
):
163
171
.
2
Galluzzi
L,
Senovilla
L,
Vitale
I,
et al.
Molecular mechanisms of cisplatin resistance
.
Oncogene
.
2012
;
31
(
15
):
1869
1883
.
3
Vilmar
A,
Sorensen
JB.
Excision repair cross-complementation group 1 (ERCC1) in platinum-based treatment of non–small cell lung cancer with special emphasis on carboplatin: a review of current literature
.
Lung Cancer
.
2009
;
64
(
2
):
131
139
.
4
Smith
DH,
Fiehn
AM,
Fogh
L,
et al.
Measuring ERCC1 protein expression in cancer specimens: validation of a novel antibody
.
Sci Rep
.
2014
;
4
:
4313
.
5
Olaussen
KA,
Dunant
A,
Fouret
P,
et al
IALT Bio Investigators. DNA repair by ERCC1 in non–small cell lung cancer and cisplatin-based adjuvant chemotherapy
.
N Engl J Med
.
2006
;
355
(
10
):
983
991
.
6
Friboulet
L,
Olaussen
KA,
Pignon
JP,
et al.
ERCC1 isoform expression and DNA repair in non–small cell lung cancer
.
N Engl J Med
.
2013
;
368
(
12
):
1101
1110
.
7
Bauman
JE,
Austin
MC,
Schmidt
R,
et al.
ERCC1 is a prognostic biomarker in locally advanced head and neck cancer: results from a randomised, phase II trial
.
Br J Cancer
.
2013
;
109
(
8
):
2096
2105
.
8
Lee
KH,
Min
HS,
Han
SW,
et al.
ERCC1 expression by immunohistochemistry and EGFR mutations in resected non–small cell lung cancer
.
Lung Cancer
.
2008
;
60
(
3
):
401
407
.
9
Tiseo
M,
Bordi
P,
Bortesi
B,
et al
Bio-FAST Trial Group. ERCC1/BRCA1 expression and gene polymorphisms as prognostic and predictive factors in advanced NSCLC treated with or without cisplatin
.
Br J Cancer
.
2013
;
108
(
8
):
1695
1703
.
10
Handra-Luca
A,
Hernandez
J,
Mountzios
G,
et al.
Excision repair cross complementation group 1 immunohistochemical expression predicts objective response and cancer-specific survival in patients treated by cisplatin-based induction chemotherapy for locally advanced head and neck squamous cell carcinoma
.
Clin Cancer Res
.
2007
;
13
(
13
):
3855
3859
.
11
Chiu
TJ,
Chen
CH,
Chien
CY,
Li
SH,
Tsai
HT,
Chen
YJ.
High ERCC1 expression predicts cisplatin-based chemotherapy resistance and poor outcome in unresectable squamous cell carcinoma of head and neck in a betel-chewing area
.
J Transl Med
.
2011
;
9
:
31
.
12
Fujii
T,
Toyooka
S,
Ichimura
K,
et al.
ERCC1 protein expression predicts the response of cisplatin-based neoadjuvant chemotherapy in non–small cell lung cancer
.
Lung Cancer
.
2008
;
59
(
3
):
377
384
.
13
Kim
MK,
Cho
KJ,
Kwon
GY,
et al.
ERCC1 predicting chemoradiation resistance and poor outcome in oesophageal cancer
.
Eur J Cancer
.
2008
;
44
(
1
):
54
60
.
14
Steffensen
KD,
Waldstrom
M,
Jeppesen
U,
Brandslund
I,
Jakobsen
A.
Prediction of response to chemotherapy by ERCC1 immunohistochemistry and ERCC1 polymorphism in ovarian cancer
.
Int J Gynecol Cancer
.
2008
;
18
(
4
):
702
710
.
15
Azuma
K,
Sasada
T,
Kawahara
A,
et al.
Expression of ERCC1 and class III beta-tubulin in non–small cell lung cancer patients treated with carboplatin and paclitaxel
.
Lung Cancer
.
2009
;
64
(
3
):
326
333
.
16
Ma
D,
Baruch
D,
Shu
Y,
et al.
Using protein microarray technology to screen anti-ERCC1 monoclonal antibodies for specificity and applications in pathology
.
BMC Biotechnol
.
2012
;
12
:
88
.
17
Bhagwat
NR,
Roginskaya
VY,
Acquafondata
MB,
Dhir
R,
Wood
RD,
Niedernhofer
LJ.
Immunodetection of DNA repair endonuclease ERCC1-XPF in human tissue
.
Cancer Res
.
2009
;
69
(
17
):
6831
6838
.
18
Hayes
M,
Lan
C,
Yan
J,
et al.
ERCC1 expression and outcomes in head and neck cancer treated with concurrent cisplatin and radiation
.
Anticancer Res
.
2011
;
31
(
12
):
4135
4139
.
19
Wolff
AC,
Hammond
ME,
Schwartz
JN,
et al
American Society of Clinical Oncology/College of American Pathologists. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer
.
Arch Pathol Lab Med
.
2007
;
131
(
1
):
18
43
.
20
Wolff
AC,
Hammond
ME,
Hicks
DG,
et al
American Society of Clinical Oncology; College of American Pathologists. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update
.
Arch Pathol Lab Med
.
2014
;
138
(
2
):
241
256
.
21
Fitzgibbons
PL,
Bradley
LA,
Fatheree
LA,
et al
College of American Pathologists Pathology and Laboratory Quality Center. Principles of analytic validation of immunohistochemical assays: guideline from the College of American Pathologists Pathology and Laboratory Quality Center
.
Arch Pathol Lab Med
.
2014
;
138
(
11
):
1432
1443
.
22
Center for Drug Evaluation and Research
.
Guidance For Industry: Bioanalytical Method Validation—Draft Guidance
. Revision 1. Washington, DC: US Department of Health and Human Services;
September
2013
.
23
Chen
WH,
Xin
PL,
Pan
QX,
et al.
ERCC1 single nucleotide polymorphism C8092A, but not its expression is associated with survival of esophageal squamous cell carcinoma patients from Fujian province, China
.
PloS one
.
2014
;
9
(
9
):
e106600
.

Author notes

All authors were employees of Takeda Pharmaceuticals Inc when this study was written. This study was funded by Takeda Pharmaceuticals Inc. The authors have no other relevant financial interest in the products or companies described in this article.